Acessibilidade / Reportar erro

Cytotoxic effect of combining two antisense oligonucleotides against telomerase rna component (hTR and mRNA of centromere protein B (CENP-B) in hepatocellular carcinoma cells

Abstract

Telomerase is a ribonucleoprotein enzyme that plays a crucial role in maintaining the malignancy and is responsible for cellular immortality and tumorigenesis. On another hand, Centromere protein B (CENP-B) plays an important role in cell cycle regulation and helping in the high rate proliferation of cancer cells. Our study is designed to evaluate the effect of using combined antisense oligonucleotides (ASOs) targeting (hTR) and mRNA of CENP-B on liver cancer cells. Compared with a single treatment, combination treatment with Locked Nucleic Acid (LNA) ASO (hTR) and (CENP-B) (6.25 nM from each) exhibit the maximum synergistic cytotoxic effect. hTR and CENP-B mRNA was abrogated while hTERT expression was disappeared. Caspase-3, Bax, and Bcl-2 were not detected, indicating caspase-independent cell death. A significant reduction in [Tumor necrosis factor (TNF-α) and Transforming growth factor (TGF-β)] coincides with elevation in Nitric oxide (NO) secretions was observed. Taken together; our data suggest that combination treatment with LNA ASO (hTR) and (CENP-B) could provide a promising strategy for cancer treatment by controlling many pathways concurrently. This might open a new prospective application of antisense in cancer therapy.

Key words
Telomerase; hTR; CENP-B; antisense oligonucleotide

INTRODUCTION

The telomere is a TTAGGG repeat that protects chromosomes against degradation and end-to-end fusion. Telomerase, a ribonucleoprotein enzyme (Chen et al. 2006CHEN XJ, ZHENG W, CHEN LL, CHEN ZB & WANG SQ. 2006. Telomerase antisense inhibition for the proliferation of endometrial cancer in vitro and in vivo. IJGC 16: 1987-1993. https://doi.org/10.1111/j.1525-1438.2006.00734.x.
https://doi.org/10.1111/j.1525-1438.2006...
), is responsible for the immortality of tumor cells by shielding the telomere (Asghari-Kia et al. 2017ASGHARI-KIA L, BASHASH D, SAFAROGHLI-AZAR A, MOMENY M, HAMIDPOUR M & GHAFFARI SH. 2017. Targeting human telomerase RNA component using antisense oligonucleotide induces rapid cell death and increases ATO-induced apoptosis in APL cells. Eur J Pharmacol 809: 215-223. https://doi.org/10.1016/j.ejphar.2017.05.039.
https://doi.org/10.1016/j.ejphar.2017.05...
). It has been reported that telomerase expression and activation is a crucial step for cancer cells to prevent aging (Ye et al. 2005YE J, WU YL, ZHANG S, CHEN Z, GUO LX, ZHOU RY & XIE H. 2005. Inhibitory effect of human telomerase antisense oligodeoxyribonucleotides on the growth of gastric cancer cell lines in variant tumor pathological subtype. WJG 11(15): 2230-2237. https://doi.org/10.3748/wjg.v11.i15.2230.
https://doi.org/10.3748/wjg.v11.i15.2230...
). Telomerase is made up of three main subunits; the RNA component (hTR) serves as a template for elongation of the telomere, the enzymatic active part [the telomerase reverse transcriptase (hTERT)], and telomerase-associated protein (TP1) (Yatabe et al. 2002YATABE N, KYO S, KONDO S, KANAYA T, WANG Z, MAIDA Y, TAKAKURA M, NAKAMURA M, TANAKA M & INOUE M. 2002. 2-5a Antisense Therapy Directed Against Human Telomerase Rna Inhibits Telomerase Activity and Induces Apoptosis Without Telomere Impairment in Cervical Cancer Cells. Cancer Gene Ther 9: 624-630. https://doi.org/10.1038/sj.cgt.7700479.
https://doi.org/10.1038/sj.cgt.7700479...
). Several studies have shown that although telomerase is found in most cancer cell types, it is not detected in normal cells (Chen et al. 2006CHEN XJ, ZHENG W, CHEN LL, CHEN ZB & WANG SQ. 2006. Telomerase antisense inhibition for the proliferation of endometrial cancer in vitro and in vivo. IJGC 16: 1987-1993. https://doi.org/10.1111/j.1525-1438.2006.00734.x.
https://doi.org/10.1111/j.1525-1438.2006...
). While telomeres shortened with each cell division in normal human cells, they continuously elongated in tumor cells (Yu et al. 2015YU C, YU Y, XU Z, LI H, YANG D, XIANG M, ZUO Y, LI S, CHEN Z & YU Z. 2015. ANTISENSE oligonucleotides targeting human telomerase mRNA increases the radiosensitivity of nasopharyngeal carcinoma cells. Mol Med Rep 11: 2825-2830. https://doi.org/10.3892/mmr.2014.3105.
https://doi.org/10.3892/mmr.2014.3105...
). Thus, researches focused on telomerase due to its pivotal role to be used as a selective target for new anticancer drugs (Yatabe et al. 2002YATABE N, KYO S, KONDO S, KANAYA T, WANG Z, MAIDA Y, TAKAKURA M, NAKAMURA M, TANAKA M & INOUE M. 2002. 2-5a Antisense Therapy Directed Against Human Telomerase Rna Inhibits Telomerase Activity and Induces Apoptosis Without Telomere Impairment in Cervical Cancer Cells. Cancer Gene Ther 9: 624-630. https://doi.org/10.1038/sj.cgt.7700479.
https://doi.org/10.1038/sj.cgt.7700479...
, Yu et al. 2015YU C, YU Y, XU Z, LI H, YANG D, XIANG M, ZUO Y, LI S, CHEN Z & YU Z. 2015. ANTISENSE oligonucleotides targeting human telomerase mRNA increases the radiosensitivity of nasopharyngeal carcinoma cells. Mol Med Rep 11: 2825-2830. https://doi.org/10.3892/mmr.2014.3105.
https://doi.org/10.3892/mmr.2014.3105...
).

In normal cell proliferation, microtubules attach to Centromere/Kinetochore in the segregation of paired sister chromatids during cell division (Qianjin et al. 2002QIANJIN L, QIAN W, HAIYAN LIN, SONG LUO, CHUNMEI W, DACHENG HE & YONGCHAO W. 2002. Effects of antisense CENP-B on the proliferation of HeLa (Tet-off) cells. CSB 47:379-383. doi: 10.1360/02tb9089., Tan et al. 2014TAN T, CHEN Z, LEI Y, ZHU Y & LIANG Q. 2014. A regulatory effect of INMAP on centromere proteins: Antisense INMAP induces CENP-B variation and centromeric halo. PLoS ONE 14(3): e91937. https://doi.org/10.1371/journal.pone.0091937.
https://doi.org/10.1371/journal.pone.009...
). Centromere/kinetochore complex is the area in that DNA combines with centromere proteins, which are required to stabilize the intact microtubes in bi-orientation chromosomes (Song et al. 2005SONG LUO, HAIYAN LIN, JIANGUO QI & YONGCHAO W. 2005. Effects of sense and antisense centromere/kinetochore complex protein-B (CENP-B) in cell cycle regulation. CSB 50(24): 2836-2843. doi: 10.1360/982005-910., Okada et al. 2007OKADA T, OHZEKI J, NAKANO M, YODA K, BRINKLEY WR, LARIONOV V & MASUMOTO H. 2007. CENP-B controls centromere formation depending on the chromatin context. Cell 131: 1287-300. https://doi.org/10.1016/j.cell.2007.10.045.
https://doi.org/10.1016/j.cell.2007.10.0...
). Centromere protein B (CENP-B) is 80 kD constitutive protein that passes in the inner domain of Centromere/Kinetochore complex by binding with 17 bp CENP-B box in α-satellite DNA (Song et al. 2005SONG LUO, HAIYAN LIN, JIANGUO QI & YONGCHAO W. 2005. Effects of sense and antisense centromere/kinetochore complex protein-B (CENP-B) in cell cycle regulation. CSB 50(24): 2836-2843. doi: 10.1360/982005-910.). However, CENP-B is highly expressed during cancer cell proliferation, and its down-regulation leads to a reduction in the cancer DNA replication activity and, consequently, cancer cell death (Song et al. 2005SONG LUO, HAIYAN LIN, JIANGUO QI & YONGCHAO W. 2005. Effects of sense and antisense centromere/kinetochore complex protein-B (CENP-B) in cell cycle regulation. CSB 50(24): 2836-2843. doi: 10.1360/982005-910.), the absence of CENP-B in normal cells does not markedly affect the cell division. CENP-B null mice seemed normal, and its functional centromere/kinetochore complex is maintained (Okada et al. 2007OKADA T, OHZEKI J, NAKANO M, YODA K, BRINKLEY WR, LARIONOV V & MASUMOTO H. 2007. CENP-B controls centromere formation depending on the chromatin context. Cell 131: 1287-300. https://doi.org/10.1016/j.cell.2007.10.045.
https://doi.org/10.1016/j.cell.2007.10.0...
, Tan et al. 2014TAN T, CHEN Z, LEI Y, ZHU Y & LIANG Q. 2014. A regulatory effect of INMAP on centromere proteins: Antisense INMAP induces CENP-B variation and centromeric halo. PLoS ONE 14(3): e91937. https://doi.org/10.1371/journal.pone.0091937.
https://doi.org/10.1371/journal.pone.009...
).

Antisense oligodeoxynucleotides (ASOs) are designed molecules in a sequence-specific earmarked manner to be complemented with targeted nucleic acids (RNA), this hybridization prompts call up and initiates RNase H which degrades RNA/ASO duplex (Yatabe et al. 2002YATABE N, KYO S, KONDO S, KANAYA T, WANG Z, MAIDA Y, TAKAKURA M, NAKAMURA M, TANAKA M & INOUE M. 2002. 2-5a Antisense Therapy Directed Against Human Telomerase Rna Inhibits Telomerase Activity and Induces Apoptosis Without Telomere Impairment in Cervical Cancer Cells. Cancer Gene Ther 9: 624-630. https://doi.org/10.1038/sj.cgt.7700479.
https://doi.org/10.1038/sj.cgt.7700479...
, Chen et al. 2006CHEN XJ, ZHENG W, CHEN LL, CHEN ZB & WANG SQ. 2006. Telomerase antisense inhibition for the proliferation of endometrial cancer in vitro and in vivo. IJGC 16: 1987-1993. https://doi.org/10.1111/j.1525-1438.2006.00734.x.
https://doi.org/10.1111/j.1525-1438.2006...
). Locked Nucleic Acid (LNA) GapmeR is a single-strand effective antisense agent that exhibits a high selective binding affinity to the targeted RNA (Grünweller et al. 2003GRÜNWELLER A, WYSZKO E, BIEBER B, JAHNEL R, ERDMANN VA & KURRECK J. 2003. Comparison of different antisense strategies in mammalian cells using locked nucleic acids, 2’-O-methyl RNA, phosphorothioates and small interfering RNA. Nucleic Acids Res 31: 3185-3193. https://doi.org/10.1093/nar/gkg409.
https://doi.org/10.1093/nar/gkg409...
). It is considered a promising therapeutics with a selective, potent knockdown activity (Kauppinen et al. 2005KAUPPINEN S, VESTER B & WENGEL J. 2005. Locked nucleic acid (LNA): High affinity targeting of RNA for diagnostics and therapeutics. Drug Discov Today Technol 2: 279-282. https://doi.org/10.1016/j.ddtec.2005.08.012.
https://doi.org/10.1016/j.ddtec.2005.08....
).

One of the crucial steps in cancer therapy is to develop therapeutic agents capable of silencing cancer-dependent gene(s). Antisense-based strategies against hTR have endeavored in various cancer cells, and it is more effective in blocking telomerase activity than targeting hTERT mRNA (Asghari-Kia et al. 2017ASGHARI-KIA L, BASHASH D, SAFAROGHLI-AZAR A, MOMENY M, HAMIDPOUR M & GHAFFARI SH. 2017. Targeting human telomerase RNA component using antisense oligonucleotide induces rapid cell death and increases ATO-induced apoptosis in APL cells. Eur J Pharmacol 809: 215-223. https://doi.org/10.1016/j.ejphar.2017.05.039.
https://doi.org/10.1016/j.ejphar.2017.05...
). To further improve its response, we explored the combining effect of two Antisense LNA™ GapmeR (ASOs) targeting human telomerase RNA (hTR), and mRNA of the Centromere B protein simultaneously on the liver cancer cell line (HepG2). Anti-tumor and anti-apoptotic activity of combined ASOs as well as changes in cytokine secretion levels and nitric oxide (NO) production was investigated.

MATERIALS AND METHODS

Ethical approval

All experiments were performed according to the principles of the ethics committee of the University of Sadat City. Ethics Approval No: SCU_0034722.

Oligonucleotide design and synthesis

The LNA antisense oligonucleotides (LNA ASO) against hTR with the sequence: 5’- GCTCTAGAATGAACGG- 3’ complement to the region between residues 151 and 166, LNA ASO against mRNA of CENP-B with the sequence: 5’-CACGCGGTCATCAATG-3’ complement to the region between residues 1893 and 1908 were designed and synthesized by Exiqon A/S, Denmark. Negative control LNA ASO with sequence 5’-AACACGTCTATACGC-3’ was run in parallel (Exiqon). They are nucleotides long GapmeR with phosphorothioate backbone modification.

Cell cultures

Hepatic HepG2 cancer cells (American Type Culture Collection,(ATCC) Manassas, VA) were maintained 90% confluent in a complete medium consisting of RPMI-1640 supplemented with L-glutamine (200 mM), penicillin (100 U/ml), streptomycin (100 μg/ml), sodium pyruvate 100 mM (Lonza, Basel, Switzerland), HEPES buffer (1M) and 10% heat-inactivated, mycoplasma and virus-free fetal bovine serum (FBS) at 37°C in a humidified incubator. All tissue culture chemicals purchased from Biowest (Nuaillé, France) except mentioned.

Cell viability assay

The effect of antisense oligonucleotides on tumor cells was evaluated by colorimetric 3,4,5-Dimethylthiazol-2,5-Diphenyltetrazolium bromide (MTT) assay (Sigma Aldrich, St. Louis, MO, USA). Cells were seeded in a 96-well plate to be 6000 cells/well before LNA ASOs treatment. HepG2 cells were treated with hTR LNA ASO and CENP-B LNA ASO alone and in combination in different doses (6.25nM, 12.5nM, and 25nM) using EzWay™ Transfection Reagent (Koma Biotech, Seoul, Korea) for 24 and 48 h according to manufacturer’s instructions. Cells were cultured in quadrants for each treatment. Negative control LNA ASO was run in parallel. Cell density was measured by its ability to convert tetrazolium salt to insoluble formazan crystals by mitochondrial dehydrogenase, which is solubilized by acidified Isopropanoland the absorbance was measured by ELISA reader (SunriseTM, Tecan Group. Männedorf/Switzerland) at 570 nm (Talaat et al. 2015TALAAT R, EL-SAYED W, AGWA HS, GAMAL-ELDEEN AM, MOAWIA S & ZAHRAN MAH. 2015. Anti-inflammatory effect of thalidomide dithiocarbamate and dithioate analogs. Chem Biol Interact 238: 74-81. https://doi.org/10.1016/j.cbi.2015.05.017.
https://doi.org/10.1016/j.cbi.2015.05.01...
). The relative cell viability was calculated as follows: (mean absorbance of treated cells/mean absorbance of untreated control cells) X100.

The synergistic effects and combination index (CI)

To evaluate the interaction between hTR and CENP-B ASOs, the combination index (CI) was calculated using CompuSyn Software (http://www.combosyn.com/) depending on the results of cytotoxicity after 48 hours according to the classic isobologram equation: CI=(D)1/(Dx)1+(D)2/(Dx)2, where (Dx)1 and (Dx)2 indicates the individual dose of hTR ASO and CENP-B ASO; respectively, required to inhibit a given level of viability index. (D)1 and (D)2 are the doses of hTR ASO and CENP-B ASO; respectively, necessary to produce the same effect in combination (Chou 2010CHOU TC. 2010. Drug combination studies and their synergy quantification using the chou-talalay method. Cancer Res 70: 440-446. https://doi.org/10.1158/0008-5472.CAN-09-1947.
https://doi.org/10.1158/0008-5472.CAN-09...
).

HepG2 transfection in 6-well plates

Cells were seeded in 6 well culture plates and allowed to grow on the day before transfection with ASOs. Cell confluence reached 80% after overnight culture. Transfection of HepG2 cells with different doses of LNA ASO (hTR) and LNA ASO (CENP-B) (6.25 nM. 12.5 nM, 25 nM) either alone or in combination was performed according to the instructions of EzwayTM transfection reagent. After 48 h incubation at 37ºC, supernatants were collected and stored at -80°C for the later measurement of cytokine levels, cells were trypsinized, and total RNA was extracted using TRIzolTMreagent (Invitrogen, Thermo Fisher Scientific, USA) according to the manufacturer’s instructions.

Estimation of transforming growth factor (TGF-β) and tumor necrosis factor (TNF-α)

TGF-β and TNF-α were measured in collected supernatants by Duoset enzyme-linked immunosorbent assay (ELISA) (R&D Systems Inc., Minneapolis, MN) according to the manufacturer protocol. Absorbencies were measured at 450 nm using the UV-max ELISA plate reader (SunriseTM). The ELISA reader-controlling software (Magellan, V 7.1) readily processes the digital data of raw absorbance value into a standard curve from which cytokine concentrations of unknown samples can be derived directly. Results are expressed as a pictogram of cytokine per milliliter of the supernatant.

Estimation of nitric oxide (NO)

Evaluation of NO concentration in the collected supernatant was performed using a nitric oxide assay kit (Biodiagnostic Co., Egypt) as described in the kit user guide. The assay depends on the reduction of nitrate by vanadium (III) chloride (VCl3) combined with detection by the acidic Griess reaction, which leads to bright reddish azo dye. Absorbencies were measured at 540 nm by the microplate ELISA reader (SunriseTM), and the results for Nitrite in samples were referred to nitrate standard and calculated in µmol/L (mean absorbance of sample/mean absorbance of the standard) X 50.

Apoptosis evaluation

Apoptotic gene expression was performed as previously described by (Talaat et al. 2019TALAAT RM, ABO-ZEID TM, ABO-ELFADL MT, EL-MAADAWY EA & HASSANIN MM. 2019. Combined hyperthermia and radiation therapy for treatment of hepatocellular carcinoma. APJCP 20: 2303-2310. https://doi.org/10.31557/APJCP.2019.20.8.2303.
https://doi.org/10.31557/APJCP.2019.20.8...
). In brief, extracted RNA was dissolved in RNase/DNase free water and quantified spectrophotometrically using NanoDrop 2000c spectrophotometer (Thermo Fischer Scientific, USA) and stored at - 80°C degrees. Verso one-step RT-PCR Ready Mix kit (Thermo Fisher Scientific) was used to detect the expression of desired genes; hTR, CENP-B (primers designed by primer-blast), caspase-3 (Fidan-Yaylalı et al. 2016FIDAN-YAYLALI G, DODURGA Y, SEÇME M & ELMAS L. 2016. Antidiabetic exendin-4 activates apoptotic pathway and inhibits growth of breast cancer cells. Tumor Biol 37: 2647-2653. https://doi.org/10.1007/s13277-015-4104-9.
https://doi.org/10.1007/s13277-015-4104-...
), Bax, and Bcl-2 (Huang et al. 2011HUANG TT, LIN HC, CHEN CC, LU CC, WEI CF, WU TS, LIU FG & LAI HC. 2011. Resveratrol induces apoptosis of human nasopharyngeal carcinoma cells via activation of multiple apoptotic pathways. J Cell Physiol 226: 720-728. https://doi.org/10.1002/jcp.22391.
https://doi.org/10.1002/jcp.22391...
). The effect of hTR LNA (ASO) on telomerase activity is also estimated by hTERT gene expression (Kim et al. 2016KIM JY, AN YM & PARK JH. 2016. Role of GLTSCR2 in the regulation of telomerase activity and chromosome stability. Mol Med Rep 14: 1697-1703. https://doi.org/10.3892/mmr.2016.5427.
https://doi.org/10.3892/mmr.2016.5427...
). Primer sets and PCR cycling conditions were summarized in (Table I). The PCR products were separated on agarose gel containing ethidium bromide solution (Sigma-Aldrich), and visualized by transilluminator Gel Documentation 3UV Benchtop System, and analyzed by DOC-IT (MSA) program (Ultra-Violet Products Ltd., UK). β-actin RNAs are used as the internal control for normalization (Li et al. 2016LI S ET AL. 2016. Induction of epithelial-mesenchymal transition (EMT) by Beclin 1 knockdown via posttranscriptional upregulation of ZEB1 in thyroid cancer cells. Oncotarget 7: 70364-70377. https://doi.org/10.18632/oncotarget.12217.
https://doi.org/10.18632/oncotarget.1221...
).

Table I
PCR Primers sequence and conditions.

Statistical analysis

All the statistical analyses were performed with SPSS19.0 (SPSS, Inc., Chicago, IL). Cytotoxicity data were conducted in a quadrate and repeated three times and presented as mean (x) with the corresponding relative standard deviation (RSD) [RSD = (SX100) / x̄ where S=standard deviation and x̄ = mean]. Cytokine data are expressed as mean ± standard error (x ± SE). Differences between groups were analyzed using a one-way analysis of variance of nonparametric data (Kruskal-Wallis test). Mann-Whitney U test was used as a post-hoc test. P <0.05 was considered statistically significant.

RESULTS

Evaluation of cytotoxic properties for LNA ASO against hTR and LNA ASO against CENP-B mRNAs

To assess the potential cytotoxicity of the LNA ASOs, cell viability was determined by MTT. HepG2 cells were treated individually with 6.25 nM, 12.5 nM, and 25 nM LNA ASO (hTR) or LNA ASO (CENP-B) for 24 h and 48 h (Figure 1). Compared to the control groups, reduction in cell proliferation was time and concentration-dependent (Supplementary Material - Table SI). The concentration of LNA ASO (hTR) leading to a 50% reduction in cell viability (IC50) was 38.25 nM and 26.1 nM for LNA ASO (CENP-B) after 24 h. Looking at the cytotoxic effect of LNA ASO (hTR) and LNA ASO (CENP-B) after 48 h, a severe cytotoxic effect was observed leading to less than 50% viability in all concentrations (27.47, 24.58, 18.55, and 41.2, 35.66, 26.51 for hTR and CENP-B at 6.25, 12.5, 25 nM; respectively). On the other hand, it appears that LNA ASO (hTR) has a stronger antiproliferative effect than LNA ASO (CENP-B) in all-time intervals (24 and 48 h).

Figure 1
Cytotoxic effect of individual LNA ASO hTR and LNA ASO CENP-B after 24 h and 48 h on cell viability. The experiment was repeated three times.

The combination of LNA ASO (hTR) and LNA ASO (CENP-B) examined for their cytotoxic activity on HepG2 cells (Table SII). We found that LNA ASO CENP-B could potentiate the anti-cancer property of LNA ASO (hTR) in some concentrations. As shown in (Figure 2), some combination of LNA ASO (hTR) and LNA ASO (CENP-B) was more effective in inhibiting cell viability as compared to their individual effect. The maximum inhibition in cell viability was estimated at 48 h post-treatment with LNA ASO (hTR) and LNA ASO (CENP-B) at 6.25nM combination from each.

Figure 2
Cytotoxic effects comparison of individual and in-combination LNA antisense oligonucleotides hTR ASO (T) and CENP-B ASO (B). The experiment was repeated three times. Values are given as mean ± RSD. P ≤ 0.05, (**) P ≤ 0.01, and (***) P ≤ 0.001 represent significant changes from negative control.

To investigate to what extent LNA ASO (hTR) and LNA ASO (CENP-B) act synergistically, we used Chou T.C. Combination Index by CompuSyn software (Theorem), which calculated the combination index (CI) values. CI < 1, >1, and =1 indicate synergism, antagonism, and additive effect; respectively. The fraction-affect (FA) versus log (CI) analysis revealed that all the concentration points, which are below the line of additive effects exhibited a synergistic anti-proliferative effect (Figure 3a). In agreement with the CI analysis, the isobologram analysis also demonstrated the synergistic cytotoxic effect of LNA ASO (hTR) when combined with LNA ASO (CENP-B) (Figure 3b). The synergistic and antagonist concentrations are shown in (Table SIII).

Figure 3
(a) Logarithmic Combination Index Plot and (b) Isobologram are for visual determination of synergism or antagonism calculated according to the classic isobologram equation: CI=(D)1/(Dx)1+(D)2/(Dx)2, where (Dx)1 and (Dx)2 indicates the individual dose of hTR ASO (T) and CENP-B ASO (B); respectively, required to inhibit the given level of viability index. (D)1 and (D)2 are the doses of hTR ASO and CENP-B ASO; respectively, necessary to produce the same effect in combination.

The effect of LNA ASO (hTR) and ASO LNA (CENP-B) on TGF-β and TNF-α

We assessed TGF-β and TNF-α secretion behavior 48 hours after treatment of HepG2 with LNA ASO (hTR) and LNA ASO (CENP-B) individually or combined with different concentrations. Figure 4 showed a reduction in TGF-β after treating cells with 6.25 nM, 12.5 nM, or 25 nM LNA ASO (hTR) and ASO LNA (CENP-B) either alone or in combination. The reduction was statistically significant for ASO LNA (CENP-B) alone or in combinations with LNA ASO (hTR), except for 6.25 nM LNA ASO (hTR). As compared with untreated cells, a slight, insignificant reduction was observed with all solitary concentrations of LNA ASO (hTR).

Figure 4
TGF-β and TNF-α expression levels in the supernatant after 48 h from exposure to LNA ASOs. Values are given as mean ± S.E. (*) P≤0.05 and (**) P≤0.01 represents significant changes from the negative control.

No change in TNF-α secretion was observed in 6.25 nM and 12.5 nM LNA ASO (hTR). In contrast, treating cells 25 nM of LNA ASO hTR displayed a significant (p<0.01) reduction in TNF-α. In ASO LNA (CENP-B), the elevation of TNF-αdetected followed by a significant reduction in 12.5nM and 25 nM as compared with untreated cancer cells. Considering the dual treatment of HepG2 cells with both antisenses, a reduction in TNF-α notified.

Estimation of (NO) secretion

The difference in NO secretion was evaluated in the collected supernatant 48 h after treating cancer cells with LNA-GapmeR antisense oligonucleotide either alone or combined. Our results demonstrated a significant elevation in NO level in all concentrations alone or in combinations excluding 25 nM LNA ASO (hTR) + 25 nM ASO LNA (CENP-B) (Figure 5).

Figure 5
Nitric oxide secretion level in the supernatant after 48 h from exposure to LNA ASOs. Values are given as mean ± S.E. (*) P≤0.05 and (**) P≤0.01 represents significant changes from the negative control.

Effect of LNA-GapmeR antisense oligonucleotides on RNAs

To assist the efficacy of LNA-GapmeR antisense oligonucleotides on target RNAs (hTR and mRNA (CENP-B), total RNAs extracted 48 h post-treatment were tested for the presence of hTR and mRNA CENP-B by RT-PCR. Compared with untreated cells, all concentrations of LNA-GapmeR antisense oligonucleotides (6.25 nM, 12 nM, and 25 nM) showed potent degradation for hTR, and mRNA CENP-B. Telomerase activity was measured by detection of the expression level of hTERT gene as a reflection of anticancer activity of LNA ASO (hTR), and ASO LNA (CENP-B) combined or individually. Inhibition of hTERT level after transfection with antisense (solo or combined) comparing to control HepG2 cells was found. Apoptosis evaluated by measuring the expression of Caspase-3, Bax, and Bcl-2. No detected expression for three apoptotic genes, indicating cell death via caspase-3 independent pathway by our LNA-GapmeRs. Antisense data was compared with HepG2 cells treated with the Tamoxifen drug (20µM) as a caspase-dependent apoptotic pathway (Figures S1(a,b,c,d) and S2(a, b, c)).

DISCUSSION

Antisense oligonucleotide treatment shows promising results in clinical trials. The latest accomplishments in the application of antisense oligonucleotides in clinical trials have revolutionized therapeutic methodologies for humans especially for treating cancers which require specific focuses at cell levels (Rinaldi & Wood 2017RINALDI C & WOOD MJA. 2017. Antisense oligonucleotides: the next frontier for treatment of neurological disorders. NPG 14: 9-21. https://doi.org/10.1038/nrneurol.2017.148.
https://doi.org/10.1038/nrneurol.2017.14...
, Shen & Corey 2018SHEN X & COREY DR. 2018. Chemistry, mechanism and clinical status of antisense oligonucleotides and duplex RNAs. Nucleic Acids Res 46: 1584-1600. https://doi.org/10.1093/nar/gkx1239.
https://doi.org/10.1093/nar/gkx1239...
). In this study, we aimed to evaluate the cytotoxic effect of using two LNA™ GapmeR antisense oligonucleotides (ASO) simultaneously targeting (hTR) and mRNA of CENP-B on HepG2 cancer cells.

Antisense LNA GapmeRs with phosphorothioate backbone modification were used as these molecules had the desirable characteristics of binding affinity, specificity, intracellular penetration, and stability compared to different antisense techniques (Grünweller et al. 2003GRÜNWELLER A, WYSZKO E, BIEBER B, JAHNEL R, ERDMANN VA & KURRECK J. 2003. Comparison of different antisense strategies in mammalian cells using locked nucleic acids, 2’-O-methyl RNA, phosphorothioates and small interfering RNA. Nucleic Acids Res 31: 3185-3193. https://doi.org/10.1093/nar/gkg409.
https://doi.org/10.1093/nar/gkg409...
, Kauppinen et al. 2005KAUPPINEN S, VESTER B & WENGEL J. 2005. Locked nucleic acid (LNA): High affinity targeting of RNA for diagnostics and therapeutics. Drug Discov Today Technol 2: 279-282. https://doi.org/10.1016/j.ddtec.2005.08.012.
https://doi.org/10.1016/j.ddtec.2005.08....
). In our study, LNA ASO against hTR is used to get the maximum selectivity and efficacy to inhibit telomerase activity. The results obtained from previous studies showed that targeting the hTR is more direct and efficient than targeting hTERT mRNA or hTERT protein (Natarajan et al. 2004NATARAJAN S, CHEN Z, WANCEWICZ EV, MONIA BP & COREY DR. 2004. Telomerase reverse transcriptase (hTERT) mRNA and telomerase RNA (hTR) as targets for downregulation of telomerase activity. Oligonucleotides 14: 263-273. https://doi.org/10.1089/oli.2004.14.263.
https://doi.org/10.1089/oli.2004.14.263...
, Li et al. 2011LI Y ET AL. 2011. Telomerase inhibition strategies by siRNAs against either hTR or hTERT in oral squamous cell carcinoma. Cancer Gene Ther 18: 318-325. https://doi.org/10.1038/cgt.2010.81
https://doi.org/10.1038/cgt.2010.81 ...
). Our data revealed that treating HepG2 cells with LNA ASO (hTR) reduces its survival rate and induces time- and concentration-dependent cytotoxic effects. These results are consistent with several documented studies (Koga et al. 2001KOGA S, KONDO Y, KOMATA T & KONDO S. 2001. Treatment of bladder cancer cells in vitro and in vivo with 2-5A antisense telomerase RNA. Gene Ther 8: 654-658. https://doi.org/10.1038/sj.gt.3301449.
https://doi.org/10.1038/sj.gt.3301449...
, Yatabe et al. 2002YATABE N, KYO S, KONDO S, KANAYA T, WANG Z, MAIDA Y, TAKAKURA M, NAKAMURA M, TANAKA M & INOUE M. 2002. 2-5a Antisense Therapy Directed Against Human Telomerase Rna Inhibits Telomerase Activity and Induces Apoptosis Without Telomere Impairment in Cervical Cancer Cells. Cancer Gene Ther 9: 624-630. https://doi.org/10.1038/sj.cgt.7700479.
https://doi.org/10.1038/sj.cgt.7700479...
, Asghari-Kia et al. 2017ASGHARI-KIA L, BASHASH D, SAFAROGHLI-AZAR A, MOMENY M, HAMIDPOUR M & GHAFFARI SH. 2017. Targeting human telomerase RNA component using antisense oligonucleotide induces rapid cell death and increases ATO-induced apoptosis in APL cells. Eur J Pharmacol 809: 215-223. https://doi.org/10.1016/j.ejphar.2017.05.039.
https://doi.org/10.1016/j.ejphar.2017.05...
). Of note, we have demonstrated that LNA ASO (hTR) completely inhibits telomerase enzyme and diminish hTERT mRNA and hTR effectively.

CENP-B has a significant control effect on kinetochore composition and the centromeres function (Okada et al. 2007OKADA T, OHZEKI J, NAKANO M, YODA K, BRINKLEY WR, LARIONOV V & MASUMOTO H. 2007. CENP-B controls centromere formation depending on the chromatin context. Cell 131: 1287-300. https://doi.org/10.1016/j.cell.2007.10.045.
https://doi.org/10.1016/j.cell.2007.10.0...
, Fachinetti et al. 2015FACHINETTI D, HAN JS, MCMAHON MA, LY P, ABDULLAH A, WONG AJ & CLEVELAND DW. 2015. DNA Sequence-Specific Binding of CENP-B Enhances the Fidelity of Human Centromere Function. Dev Cell 33(3): 314-327. https://doi.org/10.1016/j.devcel.2015.03.020.
https://doi.org/10.1016/j.devcel.2015.03...
). Interestingly, knockout of CENP-B was reported to reduce the cancer cell proliferation by lowering the progress of the cell cycle, while the cancer cell proliferation indexes back to normal after the effect of antisense CENP-B ended (Qianjin et al. 2002QIANJIN L, QIAN W, HAIYAN LIN, SONG LUO, CHUNMEI W, DACHENG HE & YONGCHAO W. 2002. Effects of antisense CENP-B on the proliferation of HeLa (Tet-off) cells. CSB 47:379-383. doi: 10.1360/02tb9089., Song et al. 2005SONG LUO, HAIYAN LIN, JIANGUO QI & YONGCHAO W. 2005. Effects of sense and antisense centromere/kinetochore complex protein-B (CENP-B) in cell cycle regulation. CSB 50(24): 2836-2843. doi: 10.1360/982005-910., Tan et al. 2014TAN T, CHEN Z, LEI Y, ZHU Y & LIANG Q. 2014. A regulatory effect of INMAP on centromere proteins: Antisense INMAP induces CENP-B variation and centromeric halo. PLoS ONE 14(3): e91937. https://doi.org/10.1371/journal.pone.0091937.
https://doi.org/10.1371/journal.pone.009...
). In the agreement, we demonstrated that LNA ASO (CENP-B) induced a cytotoxic effect, in time- and dose-dependent, on the viability of HepG2 cells. Notably, LNA ASO (CENP-B) was active in abrogating CENP-B mRNA.

Previous efforts have tried to increase the potency of chemotherapy or radiation by combining it with anti-telomerase agents. They indicated that inhibition of telomerase could potentiate cytotoxicity and decrease the side effects of chemotherapeutic agents (Berardinelli et al. 2017BERARDINELLI F, COLUZZI E, SGURA A & ANTOCCIA A. 2017. Targeting telomerase and telomeres to enhance ionizing radiation effects in in vitro and in vivo cancer models. Mutat Res Rev 773: 204-219. https://doi.org/10.1016/j.mrrev.2017.02.004.
https://doi.org/10.1016/j.mrrev.2017.02....
, Barczak et al. 2018BARCZAK W, SOBECKA A, GOLUSINSKI P, MASTERNAK MM, RUBIS B, SUCHORSKA WM & GOLUSINSKI W. 2018. HTERT gene knockdown enhances response to radio- and chemotherapy in head and neck cancer cell lines through a DNA damage pathway modification. Sci Rep 8: 1-16. https://doi.org/10.1038/s41598-018-24503-y.
https://doi.org/10.1038/s41598-018-24503...
). It is interesting to discover the impact of treating cells with more than one ASOs targeting cancer genes at the same time. Surprisingly, targeting HepG2 cells with both LNA ASO (hTR) and (CENP-B) in combinations exhibited a different cytotoxic effect manner compared to each one individually. A synergistic cytotoxic effect in low concentration combinations has been demonstrated while the impact is reversely antagonist by increasing the concentrations. Also, the cells have undergone caspase-independent apoptosis after treatment with LNA ASO (hTR) or (CENP-B) individually and in combination.

Trying to get an idea about changes in some mediators released by HepG2 in response to ASOs, we have measured TGF-β, TNF-α and NO which play an important role in cancer apoptosis, proliferation, and metastasis (Fukumura et al. 2006FUKUMURA D, KASHIWAGI S & JAIN RK. 2006. The role of nitric oxide in tumour progression. Nat Rev Cancer 6: 521-534. https://doi.org/10.1038/nrc1910.
https://doi.org/10.1038/nrc1910...
, Liu et al. 2017LIU S, CHEN S & ZENG J. 2017. TGF β signaling: A complex role in tumorigenesis (Review). Mol Med Rep 17: 699-704. https://doi.org/10.3892/mmr.2017.7970.
https://doi.org/10.3892/mmr.2017.7970...
). As increasing the level of TGF-β and TNF-α mediate metastasis, several approaches were trying to block their pathways have been conducted (Sasi et al. 2012SASI SP ET AL. 2012. Breaking the harmony of TNF-α signaling for cancer treatment. Oncogene 31: 4117-4127. https://doi.org/10.1038/onc.2011.567.
https://doi.org/10.1038/onc.2011.567...
, Neuzillet et al. 2015NEUZILLET C, TIJERAS-RABALLAND A, COHEN R, CROS J, FAIVRE S, RAYMOND E & DE GRAMONT A. 2015. Targeting the TGFβ pathway for cancer therapy. Pharmacol Ther 147: 22-31. https://doi.org/10.1016/j.pharmthera.2014.11.001.
https://doi.org/10.1016/j.pharmthera.201...
, de Gramont et al. 2017DE GRAMONT A, FAIVRE S & RAYMOND E. 2017. Novel TGF-β inhibitors ready for prime time in onco-immunology. OncoImmunology 6: 1-5. https://doi.org/10.1080/2162402X.2016.125745.
https://doi.org/10.1080/2162402X.2016.12...
, Wu et al. 2017WU X ET AL. 2017. TNF-α sensitizes chemotherapy and radiotherapy against breast cancer cells. Cancer Cell International 17: 1-12. https://doi.org/10.1186/s12935-017-0382-1.
https://doi.org/10.1186/s12935-017-0382-...
). We demonstrated that using LNA ASO (hTR) alone has not affected the secretion levels of TGF-β and TNF-α. On the contrary, at high concentration (25 nM), decreased TNF-α level has been observed. Interestingly, the addition of LNA ASO (CENP-B) to LNA ASO (hTR) led to a dramatic reduction in both TGF-β and TNF-α secreted by HepG2 making this combination a promising approach to target cancer cells.

Of particular interest, we found that combining both ASOs [LNA ASO (CENP-B) and LNA ASO (hTR)] elevated the level of NO secreted from HepG2 cells. NO is an important messenger that can regulate the tumor microenvironment through a variety of physiological and cellular pathways such as vasodilatation, cell proliferation, apoptosis, and cell cycle. Increasing NO production may recruit more anticancer drugs via increasing the blood supply to cancer cells (Somasundaram et al. 2018SOMASUNDARAM V ET AL. 2018. Molecular Mechanisms of Nitric Oxide in Cancer Progression, Signal Transduction, and Metabolism. ARS 30(8): 1124-1143. https://doi.org/10.1089/ars.2018.7527.
https://doi.org/10.1089/ars.2018.7527...
). NO can play two roles either tumor promoter or cancer suppressor (Fukumura et al. 2006FUKUMURA D, KASHIWAGI S & JAIN RK. 2006. The role of nitric oxide in tumour progression. Nat Rev Cancer 6: 521-534. https://doi.org/10.1038/nrc1910.
https://doi.org/10.1038/nrc1910...
). The role of NO as a potent anticancer agent is also documented in some reports (Huerta 2015).

CONCLUSION

Taken together, our results elucidate that a combination of ASOs could induce different patterns of cytotoxic effects than individual ones. LNA ASO (CENP-B) could potentiate the anti-cancer effect of LNA ASO (hTR) in a caspase-independent mechanism. Co-treatment of cancer cells by LNA ASO (CENP-B) and LNA ASO (hTR) has displayed a significant reduction in TGF-β and TNF-α coincides with an elevation of NO. Overall, the outcome data brace our hypothesis that combined LNA ASO (CENP-B) with LNA ASO (hTR) could augment the anti-tumor effect and provide a novel promising strategy for cancer treatment. Further investigations, including in vivo studies, are recommended to improve our results.

REFERENCES

  • ASGHARI-KIA L, BASHASH D, SAFAROGHLI-AZAR A, MOMENY M, HAMIDPOUR M & GHAFFARI SH. 2017. Targeting human telomerase RNA component using antisense oligonucleotide induces rapid cell death and increases ATO-induced apoptosis in APL cells. Eur J Pharmacol 809: 215-223. https://doi.org/10.1016/j.ejphar.2017.05.039.
    » https://doi.org/10.1016/j.ejphar.2017.05.039
  • BARCZAK W, SOBECKA A, GOLUSINSKI P, MASTERNAK MM, RUBIS B, SUCHORSKA WM & GOLUSINSKI W. 2018. HTERT gene knockdown enhances response to radio- and chemotherapy in head and neck cancer cell lines through a DNA damage pathway modification. Sci Rep 8: 1-16. https://doi.org/10.1038/s41598-018-24503-y.
    » https://doi.org/10.1038/s41598-018-24503-y
  • BERARDINELLI F, COLUZZI E, SGURA A & ANTOCCIA A. 2017. Targeting telomerase and telomeres to enhance ionizing radiation effects in in vitro and in vivo cancer models. Mutat Res Rev 773: 204-219. https://doi.org/10.1016/j.mrrev.2017.02.004.
    » https://doi.org/10.1016/j.mrrev.2017.02.004
  • CHEN XJ, ZHENG W, CHEN LL, CHEN ZB & WANG SQ. 2006. Telomerase antisense inhibition for the proliferation of endometrial cancer in vitro and in vivo. IJGC 16: 1987-1993. https://doi.org/10.1111/j.1525-1438.2006.00734.x.
    » https://doi.org/10.1111/j.1525-1438.2006.00734.x
  • CHOU TC. 2010. Drug combination studies and their synergy quantification using the chou-talalay method. Cancer Res 70: 440-446. https://doi.org/10.1158/0008-5472.CAN-09-1947.
    » https://doi.org/10.1158/0008-5472.CAN-09-1947
  • DE GRAMONT A, FAIVRE S & RAYMOND E. 2017. Novel TGF-β inhibitors ready for prime time in onco-immunology. OncoImmunology 6: 1-5. https://doi.org/10.1080/2162402X.2016.125745.
    » https://doi.org/10.1080/2162402X.2016.125745
  • FACHINETTI D, HAN JS, MCMAHON MA, LY P, ABDULLAH A, WONG AJ & CLEVELAND DW. 2015. DNA Sequence-Specific Binding of CENP-B Enhances the Fidelity of Human Centromere Function. Dev Cell 33(3): 314-327. https://doi.org/10.1016/j.devcel.2015.03.020.
    » https://doi.org/10.1016/j.devcel.2015.03.020
  • FIDAN-YAYLALI G, DODURGA Y, SEÇME M & ELMAS L. 2016. Antidiabetic exendin-4 activates apoptotic pathway and inhibits growth of breast cancer cells. Tumor Biol 37: 2647-2653. https://doi.org/10.1007/s13277-015-4104-9.
    » https://doi.org/10.1007/s13277-015-4104-9
  • FUKUMURA D, KASHIWAGI S & JAIN RK. 2006. The role of nitric oxide in tumour progression. Nat Rev Cancer 6: 521-534. https://doi.org/10.1038/nrc1910.
    » https://doi.org/10.1038/nrc1910
  • GRÜNWELLER A, WYSZKO E, BIEBER B, JAHNEL R, ERDMANN VA & KURRECK J. 2003. Comparison of different antisense strategies in mammalian cells using locked nucleic acids, 2’-O-methyl RNA, phosphorothioates and small interfering RNA. Nucleic Acids Res 31: 3185-3193. https://doi.org/10.1093/nar/gkg409.
    » https://doi.org/10.1093/nar/gkg409
  • HUANG TT, LIN HC, CHEN CC, LU CC, WEI CF, WU TS, LIU FG & LAI HC. 2011. Resveratrol induces apoptosis of human nasopharyngeal carcinoma cells via activation of multiple apoptotic pathways. J Cell Physiol 226: 720-728. https://doi.org/10.1002/jcp.22391.
    » https://doi.org/10.1002/jcp.22391
  • KAUPPINEN S, VESTER B & WENGEL J. 2005. Locked nucleic acid (LNA): High affinity targeting of RNA for diagnostics and therapeutics. Drug Discov Today Technol 2: 279-282. https://doi.org/10.1016/j.ddtec.2005.08.012.
    » https://doi.org/10.1016/j.ddtec.2005.08.012
  • KIM JY, AN YM & PARK JH. 2016. Role of GLTSCR2 in the regulation of telomerase activity and chromosome stability. Mol Med Rep 14: 1697-1703. https://doi.org/10.3892/mmr.2016.5427.
    » https://doi.org/10.3892/mmr.2016.5427
  • KOGA S, KONDO Y, KOMATA T & KONDO S. 2001. Treatment of bladder cancer cells in vitro and in vivo with 2-5A antisense telomerase RNA. Gene Ther 8: 654-658. https://doi.org/10.1038/sj.gt.3301449.
    » https://doi.org/10.1038/sj.gt.3301449
  • LI S ET AL. 2016. Induction of epithelial-mesenchymal transition (EMT) by Beclin 1 knockdown via posttranscriptional upregulation of ZEB1 in thyroid cancer cells. Oncotarget 7: 70364-70377. https://doi.org/10.18632/oncotarget.12217.
    » https://doi.org/10.18632/oncotarget.12217
  • LI Y ET AL. 2011. Telomerase inhibition strategies by siRNAs against either hTR or hTERT in oral squamous cell carcinoma. Cancer Gene Ther 18: 318-325. https://doi.org/10.1038/cgt.2010.81
    » https://doi.org/10.1038/cgt.2010.81
  • LIU S, CHEN S & ZENG J. 2017. TGF β signaling: A complex role in tumorigenesis (Review). Mol Med Rep 17: 699-704. https://doi.org/10.3892/mmr.2017.7970.
    » https://doi.org/10.3892/mmr.2017.7970
  • NATARAJAN S, CHEN Z, WANCEWICZ EV, MONIA BP & COREY DR. 2004. Telomerase reverse transcriptase (hTERT) mRNA and telomerase RNA (hTR) as targets for downregulation of telomerase activity. Oligonucleotides 14: 263-273. https://doi.org/10.1089/oli.2004.14.263.
    » https://doi.org/10.1089/oli.2004.14.263
  • NEUZILLET C, TIJERAS-RABALLAND A, COHEN R, CROS J, FAIVRE S, RAYMOND E & DE GRAMONT A. 2015. Targeting the TGFβ pathway for cancer therapy. Pharmacol Ther 147: 22-31. https://doi.org/10.1016/j.pharmthera.2014.11.001.
    » https://doi.org/10.1016/j.pharmthera.2014.11.001
  • OKADA T, OHZEKI J, NAKANO M, YODA K, BRINKLEY WR, LARIONOV V & MASUMOTO H. 2007. CENP-B controls centromere formation depending on the chromatin context. Cell 131: 1287-300. https://doi.org/10.1016/j.cell.2007.10.045.
    » https://doi.org/10.1016/j.cell.2007.10.045
  • QIANJIN L, QIAN W, HAIYAN LIN, SONG LUO, CHUNMEI W, DACHENG HE & YONGCHAO W. 2002. Effects of antisense CENP-B on the proliferation of HeLa (Tet-off) cells. CSB 47:379-383. doi: 10.1360/02tb9089.
  • RINALDI C & WOOD MJA. 2017. Antisense oligonucleotides: the next frontier for treatment of neurological disorders. NPG 14: 9-21. https://doi.org/10.1038/nrneurol.2017.148.
    » https://doi.org/10.1038/nrneurol.2017.148
  • SASI SP ET AL. 2012. Breaking the harmony of TNF-α signaling for cancer treatment. Oncogene 31: 4117-4127. https://doi.org/10.1038/onc.2011.567.
    » https://doi.org/10.1038/onc.2011.567
  • SHEN X & COREY DR. 2018. Chemistry, mechanism and clinical status of antisense oligonucleotides and duplex RNAs. Nucleic Acids Res 46: 1584-1600. https://doi.org/10.1093/nar/gkx1239.
    » https://doi.org/10.1093/nar/gkx1239
  • SOMASUNDARAM V ET AL. 2018. Molecular Mechanisms of Nitric Oxide in Cancer Progression, Signal Transduction, and Metabolism. ARS 30(8): 1124-1143. https://doi.org/10.1089/ars.2018.7527.
    » https://doi.org/10.1089/ars.2018.7527
  • SONG LUO, HAIYAN LIN, JIANGUO QI & YONGCHAO W. 2005. Effects of sense and antisense centromere/kinetochore complex protein-B (CENP-B) in cell cycle regulation. CSB 50(24): 2836-2843. doi: 10.1360/982005-910.
  • TALAAT R, EL-SAYED W, AGWA HS, GAMAL-ELDEEN AM, MOAWIA S & ZAHRAN MAH. 2015. Anti-inflammatory effect of thalidomide dithiocarbamate and dithioate analogs. Chem Biol Interact 238: 74-81. https://doi.org/10.1016/j.cbi.2015.05.017.
    » https://doi.org/10.1016/j.cbi.2015.05.017
  • TALAAT RM, ABO-ZEID TM, ABO-ELFADL MT, EL-MAADAWY EA & HASSANIN MM. 2019. Combined hyperthermia and radiation therapy for treatment of hepatocellular carcinoma. APJCP 20: 2303-2310. https://doi.org/10.31557/APJCP.2019.20.8.2303.
    » https://doi.org/10.31557/APJCP.2019.20.8.2303
  • TAN T, CHEN Z, LEI Y, ZHU Y & LIANG Q. 2014. A regulatory effect of INMAP on centromere proteins: Antisense INMAP induces CENP-B variation and centromeric halo. PLoS ONE 14(3): e91937. https://doi.org/10.1371/journal.pone.0091937.
    » https://doi.org/10.1371/journal.pone.0091937
  • WU X ET AL. 2017. TNF-α sensitizes chemotherapy and radiotherapy against breast cancer cells. Cancer Cell International 17: 1-12. https://doi.org/10.1186/s12935-017-0382-1.
    » https://doi.org/10.1186/s12935-017-0382-1
  • YATABE N, KYO S, KONDO S, KANAYA T, WANG Z, MAIDA Y, TAKAKURA M, NAKAMURA M, TANAKA M & INOUE M. 2002. 2-5a Antisense Therapy Directed Against Human Telomerase Rna Inhibits Telomerase Activity and Induces Apoptosis Without Telomere Impairment in Cervical Cancer Cells. Cancer Gene Ther 9: 624-630. https://doi.org/10.1038/sj.cgt.7700479.
    » https://doi.org/10.1038/sj.cgt.7700479
  • YE J, WU YL, ZHANG S, CHEN Z, GUO LX, ZHOU RY & XIE H. 2005. Inhibitory effect of human telomerase antisense oligodeoxyribonucleotides on the growth of gastric cancer cell lines in variant tumor pathological subtype. WJG 11(15): 2230-2237. https://doi.org/10.3748/wjg.v11.i15.2230.
    » https://doi.org/10.3748/wjg.v11.i15.2230
  • YU C, YU Y, XU Z, LI H, YANG D, XIANG M, ZUO Y, LI S, CHEN Z & YU Z. 2015. ANTISENSE oligonucleotides targeting human telomerase mRNA increases the radiosensitivity of nasopharyngeal carcinoma cells. Mol Med Rep 11: 2825-2830. https://doi.org/10.3892/mmr.2014.3105.
    » https://doi.org/10.3892/mmr.2014.3105

Publication Dates

  • Publication in this collection
    22 July 2022
  • Date of issue
    2022

History

  • Received
    17 Apr 2020
  • Accepted
    03 July 2020
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br